Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 326
Filter
Add more filters

Publication year range
1.
Cell ; 185(3): 493-512.e25, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35032429

ABSTRACT

Severe COVID-19 is linked to both dysfunctional immune response and unrestrained immunopathology, and it remains unclear whether T cells contribute to disease pathology. Here, we combined single-cell transcriptomics and single-cell proteomics with mechanistic studies to assess pathogenic T cell functions and inducing signals. We identified highly activated CD16+ T cells with increased cytotoxic functions in severe COVID-19. CD16 expression enabled immune-complex-mediated, T cell receptor-independent degranulation and cytotoxicity not found in other diseases. CD16+ T cells from COVID-19 patients promoted microvascular endothelial cell injury and release of neutrophil and monocyte chemoattractants. CD16+ T cell clones persisted beyond acute disease maintaining their cytotoxic phenotype. Increased generation of C3a in severe COVID-19 induced activated CD16+ cytotoxic T cells. Proportions of activated CD16+ T cells and plasma levels of complement proteins upstream of C3a were associated with fatal outcome of COVID-19, supporting a pathological role of exacerbated cytotoxicity and complement activation in COVID-19.


Subject(s)
COVID-19/immunology , COVID-19/pathology , Complement Activation , Proteome , SARS-CoV-2/immunology , T-Lymphocytes, Cytotoxic/immunology , Transcriptome , Adult , Aged , Aged, 80 and over , COVID-19/virology , Chemotactic Factors/metabolism , Cytotoxicity, Immunologic , Endothelial Cells/virology , Female , Humans , Lymphocyte Activation , Male , Microvessels/virology , Middle Aged , Monocytes/metabolism , Neutrophils/metabolism , Receptors, IgG/metabolism , Single-Cell Analysis , Young Adult
2.
Nat Immunol ; 22(2): 229-239, 2021 02.
Article in English | MEDLINE | ID: mdl-33398179

ABSTRACT

In chronic hepatitis C virus (HCV) infection, exhausted HCV-specific CD8+ T cells comprise memory-like and terminally exhausted subsets. However, little is known about the molecular profile and fate of these two subsets after the elimination of chronic antigen stimulation by direct-acting antiviral (DAA) therapy. Here, we report a progenitor-progeny relationship between memory-like and terminally exhausted HCV-specific CD8+ T cells via an intermediate subset. Single-cell transcriptomics implicated that memory-like cells are maintained and terminally exhausted cells are lost after DAA-mediated cure, resulting in a memory polarization of the overall HCV-specific CD8+ T cell response. However, an exhausted core signature of memory-like CD8+ T cells was still detectable, including, to a smaller extent, in HCV-specific CD8+ T cells targeting variant epitopes. These results identify a molecular signature of T cell exhaustion that is maintained as a chronic scar in HCV-specific CD8+ T cells even after the cessation of chronic antigen stimulation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hepacivirus/immunology , Hepatitis C, Chronic/immunology , Immunologic Memory/genetics , Transcriptome , Antigens, Viral/immunology , Antiviral Agents/therapeutic use , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Gene Expression Profiling , Gene Regulatory Networks , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/virology , Host-Pathogen Interactions , Humans , Phenotype , Remission Induction , Single-Cell Analysis , Treatment Outcome
3.
Cell ; 155(3): 567-81, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24139898

ABSTRACT

Mutation is a fundamental process in tumorigenesis. However, the degree to which the rate of somatic mutation varies across the human genome and the mechanistic basis underlying this variation remain to be fully elucidated. Here, we performed a cross-cancer comparison of 402 whole genomes comprising a diverse set of childhood and adult tumors, including both solid and hematopoietic malignancies. Surprisingly, we found that the inactive X chromosome of many female cancer genomes accumulates on average twice and up to four times as many somatic mutations per megabase, as compared to the individual autosomes. Whole-genome sequencing of clonally expanded hematopoietic stem/progenitor cells (HSPCs) from healthy individuals and a premalignant myelodysplastic syndrome (MDS) sample revealed no X chromosome hypermutation. Our data suggest that hypermutation of the inactive X chromosome is an early and frequent feature of tumorigenesis resulting from DNA replication stress in aberrantly proliferating cells.


Subject(s)
Chromosomes, Human, X , Mutation , Neoplasms/genetics , X Chromosome Inactivation , Adult , Aged , DNA Replication , Female , Humans , Male , Medulloblastoma/genetics , Medulloblastoma/pathology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Polymorphism, Single Nucleotide , S Phase
4.
Cell ; 148(1-2): 59-71, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22265402

ABSTRACT

Genomic rearrangements are thought to occur progressively during tumor development. Recent findings, however, suggest an alternative mechanism, involving massive chromosome rearrangements in a one-step catastrophic event termed chromothripsis. We report the whole-genome sequencing-based analysis of a Sonic-Hedgehog medulloblastoma (SHH-MB) brain tumor from a patient with a germline TP53 mutation (Li-Fraumeni syndrome), uncovering massive, complex chromosome rearrangements. Integrating TP53 status with microarray and deep sequencing-based DNA rearrangement data in additional patients reveals a striking association between TP53 mutation and chromothripsis in SHH-MBs. Analysis of additional tumor entities substantiates a link between TP53 mutation and chromothripsis, and indicates a context-specific role for p53 in catastrophic DNA rearrangements. Among these, we observed a strong association between somatic TP53 mutations and chromothripsis in acute myeloid leukemia. These findings connect p53 status and chromothripsis in specific tumor types, providing a genetic basis for understanding particularly aggressive subtypes of cancer.


Subject(s)
Brain Neoplasms/genetics , Gene Rearrangement , Medulloblastoma/genetics , Tumor Suppressor Protein p53/genetics , Animals , Child , Chromosome Aberrations , DNA Copy Number Variations , DNA Mutational Analysis , Disease Models, Animal , Humans , Leukemia, Myeloid, Acute/genetics , Li-Fraumeni Syndrome/physiopathology , Mice , Middle Aged
5.
Nature ; 599(7884): 283-289, 2021 11.
Article in English | MEDLINE | ID: mdl-34517409

ABSTRACT

Derailed cytokine and immune cell networks account for the organ damage and the clinical severity of COVID-19 (refs. 1-4). Here we show that SARS-CoV-2, like other viruses, evokes cellular senescence as a primary stress response in infected cells. Virus-induced senescence (VIS) is indistinguishable from other forms of cellular senescence and is accompanied by a senescence-associated secretory phenotype (SASP), which comprises pro-inflammatory cytokines, extracellular-matrix-active factors and pro-coagulatory mediators5-7. Patients with COVID-19 displayed markers of senescence in their airway mucosa in situ and increased serum levels of SASP factors. In vitro assays demonstrated macrophage activation with SASP-reminiscent secretion, complement lysis and SASP-amplifying secondary senescence of endothelial cells, which mirrored hallmark features of COVID-19 such as macrophage and neutrophil infiltration, endothelial damage and widespread thrombosis in affected lung tissue1,8,9. Moreover, supernatant from VIS cells, including SARS-CoV-2-induced senescence, induced neutrophil extracellular trap formation and activation of platelets and the clotting cascade. Senolytics such as navitoclax and a combination of dasatinib plus quercetin selectively eliminated VIS cells, mitigated COVID-19-reminiscent lung disease and reduced inflammation in SARS-CoV-2-infected hamsters and mice. Our findings mark VIS as a pathogenic trigger of COVID-19-related cytokine escalation and organ damage, and suggest that senolytic targeting of virus-infected cells is a treatment option against SARS-CoV-2 and perhaps other viral infections.


Subject(s)
COVID-19 Drug Treatment , COVID-19/pathology , COVID-19/virology , Cellular Senescence/drug effects , Molecular Targeted Therapy , SARS-CoV-2/pathogenicity , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , COVID-19/complications , Cell Line , Cricetinae , Dasatinib/pharmacology , Dasatinib/therapeutic use , Disease Models, Animal , Female , Humans , Male , Mice , Quercetin/pharmacology , Quercetin/therapeutic use , SARS-CoV-2/drug effects , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Thrombosis/complications , Thrombosis/immunology , Thrombosis/metabolism
6.
EMBO Rep ; 24(12): e57912, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37818799

ABSTRACT

The risk of developing severe COVID-19 rises dramatically with age. Schoolchildren are significantly less likely than older people to die from SARS-CoV-2 infection, but the molecular mechanisms underlying this age-dependence are unknown. In primary infections, innate immunity is critical due to the lack of immune memory. Children, in particular, have a significantly stronger interferon response due to a primed state of their airway epithelium. In single-cell transcriptomes of nasal turbinates, we find increased frequencies of immune cells and stronger cytokine-mediated interactions with epithelial cells, resulting in increased epithelial expression of viral sensors (RIG-I, MDA5) via IRF1. In vitro, adolescent peripheral blood mononuclear cells produce more cytokines, priming A549 cells for stronger interferon responses to SARS-CoV-2. Taken together, our findings suggest that increased numbers of immune cells in the airways of children and enhanced cytokine-based interactions with epithelial cells tune the setpoint of the epithelial antiviral system. Our findings shed light on the molecular basis of children's remarkable resistance to COVID-19 and may suggest a novel concept for immunoprophylactic treatments.


Subject(s)
COVID-19 , SARS-CoV-2 , Child , Adolescent , Humans , Aged , Leukocytes, Mononuclear , Epithelial Cells , Interferons , Immunity, Innate , Cytokines , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use
7.
Am J Respir Crit Care Med ; 209(11): 1338-1350, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38259174

ABSTRACT

Rationale: Pharmacological improvement of cystic fibrosis transmembrane conductance regulator (CFTR) function with elexacaftor/tezacaftor/ivacaftor (ETI) provides unprecedented improvements in lung function and other clinical outcomes in patients with cystic fibrosis (CF). However, ETI effects on impaired mucosal homeostasis and host defense at the molecular and cellular levels in the airways of patients with CF remain unknown. Objectives: To investigate effects of ETI on the transcriptome of nasal epithelial and immune cells from children with CF at the single-cell level. Methods: Nasal swabs from 13 children with CF and at least one F508del allele aged 6 to 11 years were collected at baseline and 3 months after initiation of ETI, subjected to single-cell RNA sequencing, and compared with swabs from 12 age-matched healthy children. Measurements and Main Results: Proportions of CFTR-positive cells were decreased in epithelial basal, club, and goblet cells, but not in ionocytes, from children with CF at baseline and were restored by ETI therapy to nearly healthy levels. Single-cell transcriptomics revealed an impaired IFN signaling and reduced expression of major histocompatibility complex classes I and II encoding genes in epithelial cells of children with CF at baseline, which was partially restored by ETI. In addition, ETI therapy markedly reduced the inflammatory phenotype of immune cells, particularly of neutrophils and macrophages. Conclusions: Pharmacological improvement of CFTR function improves innate mucosal immunity and reduces immune cell inflammatory responses in the upper airways of children with CF at the single-cell level, highlighting the potential to restore epithelial homeostasis and host defense in CF airways by early initiation of ETI therapy.


Subject(s)
Aminophenols , Benzodioxoles , Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Homeostasis , Humans , Cystic Fibrosis/drug therapy , Cystic Fibrosis/immunology , Cystic Fibrosis/physiopathology , Child , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Female , Male , Benzodioxoles/therapeutic use , Benzodioxoles/pharmacology , Aminophenols/therapeutic use , Aminophenols/pharmacology , Quinolones/therapeutic use , Quinolones/pharmacology , Indoles/therapeutic use , Indoles/pharmacology , Drug Combinations , Quinolines/therapeutic use , Quinolines/pharmacology , Pyrazoles/therapeutic use , Pyrazoles/pharmacology , Pyrroles/therapeutic use , Pyrroles/pharmacology , Nasal Mucosa/immunology , Pyridines/therapeutic use , Pyridines/pharmacology
8.
EMBO J ; 39(10): e105114, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32246845

ABSTRACT

The SARS-CoV-2 pandemic affecting the human respiratory system severely challenges public health and urgently demands for increasing our understanding of COVID-19 pathogenesis, especially host factors facilitating virus infection and replication. SARS-CoV-2 was reported to enter cells via binding to ACE2, followed by its priming by TMPRSS2. Here, we investigate ACE2 and TMPRSS2 expression levels and their distribution across cell types in lung tissue (twelve donors, 39,778 cells) and in cells derived from subsegmental bronchial branches (four donors, 17,521 cells) by single nuclei and single cell RNA sequencing, respectively. While TMPRSS2 is strongly expressed in both tissues, in the subsegmental bronchial branches ACE2 is predominantly expressed in a transient secretory cell type. Interestingly, these transiently differentiating cells show an enrichment for pathways related to RHO GTPase function and viral processes suggesting increased vulnerability for SARS-CoV-2 infection. Our data provide a rich resource for future investigations of COVID-19 infection and pathogenesis.


Subject(s)
Bronchi/cytology , Gene Expression , Lung/cytology , Peptidyl-Dipeptidase A/genetics , Serine Endopeptidases/genetics , Single-Cell Analysis , Adult , Aging , Angiotensin-Converting Enzyme 2 , Bronchi/metabolism , COVID-19 , Cells, Cultured , Chronic Disease/epidemiology , Coronavirus Infections/genetics , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Germany , Goblet Cells/metabolism , Humans , Lung/metabolism , Male , Middle Aged , Pandemics , Pneumonia, Viral/genetics , Reference Standards , Sequence Analysis, RNA , Sex Characteristics , Smoking , Tissue Banks
9.
Mol Syst Biol ; 19(7): e11799, 2023 Jul 11.
Article in English | MEDLINE | ID: mdl-37318792

ABSTRACT

In this Editorial, our Chief Editor and members of our Advisory Editorial Board discuss recent breakthroughs, current challenges, and emerging opportunities in single-cell biology and share their vision of "where the field is headed."

10.
Allergy ; 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38234010

ABSTRACT

Understanding modifiable prenatal and early life causal determinants of food allergy is important for the prevention of the disease. Randomized clinical trials studying environmental and dietary determinants of food allergy may not always be feasible. Identifying risk/protective factors for early-life food allergy often relies on observational studies, which may be affected by confounding bias. The directed acyclic graph (DAG) is a causal diagram useful to guide causal inference from observational epidemiological research. To date, research on food allergy has made little use of this promising method. We performed a literature review of existing evidence with a systematic search, synthesized 32 known risk/protective factors, and constructed a comprehensive DAG for early-life food allergy development. We present an easy-to-use online tool for researchers to re-construct, amend, and modify the DAG along with a user's guide to minimize confounding bias. We estimated that adjustment strategies in 57% of previous observational studies on modifiable factors of childhood food allergy could be improved if the researchers determined their adjustment sets by DAG. Future researchers who are interested in the causal inference of food allergy development in early life can apply the DAG to identify covariates that should and should not be controlled in observational studies.

11.
Article in German | MEDLINE | ID: mdl-38837053

ABSTRACT

The Medical Informatics Initiative (MII) funded by the Federal Ministry of Education and Research (BMBF) 2016-2027 is successfully laying the foundations for data-based medicine in Germany. As part of this funding, 51 new professorships, 21 junior research groups, and various new degree programs have been established to strengthen teaching, training, and continuing education in the field of medical informatics and to improve expertise in medical data sciences. A joint decentralized federated research data infrastructure encompassing the entire university medical center and its partners was created in the form of data integration centers (DIC) at all locations and the German Portal for Medical Research Data (FDPG) as a central access point. A modular core dataset (KDS) was defined and implemented for the secondary use of patient treatment data with consistent use of international standards (e.g., FHIR, SNOMED CT, and LOINC). An officially approved nationwide broad consent was introduced as the legal basis. The first data exports and data use projects have been carried out, embedded in an overarching usage policy and standardized contractual regulations. The further development of the MII health research data infrastructures within the cooperative framework of the Network of University Medicine (NUM) offers an excellent starting point for a German contribution to the upcoming European Health Data Space (EHDS), which opens opportunities for Germany as a medical research location.


Subject(s)
Biomedical Research , Medical Informatics , Humans , Biomedical Research/organization & administration , Germany , Health Services Research/organization & administration , Models, Organizational
12.
Brief Bioinform ; 22(6)2021 11 05.
Article in English | MEDLINE | ID: mdl-33971666

ABSTRACT

Precision oncology is a rapidly evolving interdisciplinary medical specialty. Comprehensive cancer panels are becoming increasingly available at pathology departments worldwide, creating the urgent need for scalable cancer variant annotation and molecularly informed treatment recommendations. A wealth of mainly academia-driven knowledge bases calls for software tools supporting the multi-step diagnostic process. We derive a comprehensive list of knowledge bases relevant for variant interpretation by a review of existing literature followed by a survey among medical experts from university hospitals in Germany. In addition, we review cancer variant interpretation tools, which integrate multiple knowledge bases. We categorize the knowledge bases along the diagnostic process in precision oncology and analyze programmatic access options as well as the integration of knowledge bases into software tools. The most commonly used knowledge bases provide good programmatic access options and have been integrated into a range of software tools. For the wider set of knowledge bases, access options vary across different parts of the diagnostic process. Programmatic access is limited for information regarding clinical classifications of variants and for therapy recommendations. The main issue for databases used for biological classification of pathogenic variants and pathway context information is the lack of standardized interfaces. There is no single cancer variant interpretation tool that integrates all identified knowledge bases. Specialized tools are available and need to be further developed for different steps in the diagnostic process.


Subject(s)
Databases, Genetic , Knowledge Bases , Neoplasms , Precision Medicine , Software , Humans , Neoplasms/genetics , Neoplasms/metabolism
13.
Brief Bioinform ; 22(5)2021 09 02.
Article in English | MEDLINE | ID: mdl-33589928

ABSTRACT

This article describes some use case studies and self-assessments of FAIR status of de.NBI services to illustrate the challenges and requirements for the definition of the needs of adhering to the FAIR (findable, accessible, interoperable and reusable) data principles in a large distributed bioinformatics infrastructure. We address the challenge of heterogeneity of wet lab technologies, data, metadata, software, computational workflows and the levels of implementation and monitoring of FAIR principles within the different bioinformatics sub-disciplines joint in de.NBI. On the one hand, this broad service landscape and the excellent network of experts are a strong basis for the development of useful research data management plans. On the other hand, the large number of tools and techniques maintained by distributed teams renders FAIR compliance challenging.


Subject(s)
Data Management/methods , Metadata , Neural Networks, Computer , Proteomics/methods , Software , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , International Cooperation , Phenotype , Plants/genetics , Proteome , Self-Assessment , Workflow
14.
Allergy ; 78(6): 1489-1506, 2023 06.
Article in English | MEDLINE | ID: mdl-36704932

ABSTRACT

BACKGROUND: Childhood asthma is a result of a complex interaction of genetic and environmental components causing epigenetic and immune dysregulation, airway inflammation and impaired lung function. Although different microarray based EWAS studies have been conducted, the impact of epigenetic regulation in asthma development is still widely unknown. We have therefore applied unbiased whole genome bisulfite sequencing (WGBS) to characterize global DNA-methylation profiles of asthmatic children compared to healthy controls. METHODS: Peripheral blood samples of 40 asthmatic and 42 control children aged 5-15 years from three birth cohorts were sequenced together with paired cord blood samples. Identified differentially methylated regions (DMRs) were categorized in genotype-associated, cell-type-dependent, or prenatally primed. Network analysis and subsequent natural language processing of DMR-associated genes was complemented by targeted analysis of functional translation of epigenetic regulation on the transcriptional and protein level. RESULTS: In total, 158 DMRs were identified in asthmatic children compared to controls of which 37% were related to the eosinophil content. A global hypomethylation was identified affecting predominantly enhancer regions and regulating key immune genes such as IL4, IL5RA, and EPX. These DMRs were confirmed in n = 267 samples and could be linked to aberrant gene expression. Out of the 158 DMRs identified in the established phenotype, 56 were perturbed already at birth and linked, at least in part, to prenatal influences such as tobacco smoke exposure or phthalate exposure. CONCLUSION: This is the first epigenetic study based on whole genome sequencing to identify marked dysregulation of enhancer regions as a hallmark of childhood asthma.


Subject(s)
Asthma , Epigenesis, Genetic , Female , Pregnancy , Humans , DNA Methylation , Asthma/genetics , DNA
15.
J Biomed Inform ; 144: 104437, 2023 08.
Article in English | MEDLINE | ID: mdl-37442314

ABSTRACT

BACKGROUND: The reuse of data from electronic health records (EHRs) for research purposes promises to improve the data foundation for clinical trials and may even support to enable them. Nevertheless, EHRs are characterized by both, heterogeneous structure and semantics. To standardize this data for research, the Observational Medical Outcomes Partnership (OMOP) Common Data Model (CDM) standard has recently seen an increase in use. However, the conversion of these EHRs into the OMOP CDM requires complex and resource intensive Extract Transform and Load (ETL) processes. This hampers the reuse of clinical data for research. To solve the issues of heterogeneity of EHRs and the lack of semantic precision on the care site, the openEHR standard has recently seen wider adoption. A standardized process to integrate openEHR records into the CDM potentially lowers the barriers of making EHRs accessible for research. Yet, a comprehensive approach about the integration of openEHR records into the OMOP CDM has not yet been made. METHODS: We analyzed both standards and compared their models to identify possible mappings. Based on this, we defined the necessary processes to transform openEHR records into CDM tables. We also discuss the limitation of openEHR with its unspecific demographics model and propose two possible solutions. RESULTS: We developed the OMOP Conversion Language (OMOCL) which enabled us to define a declarative openEHR archetype-to-CDM mapping language. Using OMOCL, it was possible to define a set of mappings. As a proof-of-concept, we implemented the Eos tool, which uses the OMOCL-files to successfully automatize the ETL from real-world and sample EHRs into the OMOP CDM. DISCUSSION: Both Eos and OMOCL provide a way to define generic mappings for an integration of openEHR records into OMOP. Thus, it represents a significant step towards achieving interoperability between the clinical and the research data domains. However, the transformation of openEHR data into the less expressive OMOP CDM leads to a loss of semantics.


Subject(s)
Electronic Health Records , Semantics , Databases, Factual , Reference Standards
16.
Environ Res ; 233: 116413, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37343754

ABSTRACT

While the link between exposure to high levels of ambient particulate matter (PM) and increased incidences of respiratory and cardiovascular diseases is widely recognized, recent epidemiological studies have shown that low PM concentrations are equally associated with adverse health effects. As DNA methylation is one of the main mechanisms by which cells regulate and stabilize gene expression, changes in the methylome could constitute early indicators of dysregulated signaling pathways. So far, little is known about PM-associated DNA methylation changes in the upper airways, the first point of contact between airborne pollutants and the human body. Here, we focused on cells of the upper respiratory tract and assessed their genome-wide DNA methylation pattern to explore exposure-associated early regulatory changes. Using a mobile epidemiological laboratory, nasal lavage samples were collected from a cohort of 60 adults that lived in districts with records of low (Simmerath) or moderate (Stuttgart) PM10 levels in Germany. PM10 concentrations were verified by particle measurements on the days of the sample collection and genome-wide DNA methylation was determined by enzymatic methyl sequencing at single-base resolution. We identified 231 differentially methylated regions (DMRs) between moderately and lowly PM10 exposed individuals. A high proportion of DMRs overlapped with regulatory elements, and DMR target genes were involved in pathways regulating cellular redox homeostasis and immune response. In addition, we found distinct changes in DNA methylation of the HOXA gene cluster whose methylation levels have previously been linked to air pollution exposure but also to carcinogenesis in several instances. The findings of this study suggest that regulatory changes in upper airway cells occur at PM10 levels below current European thresholds, some of which may be involved in the development of air pollution-related diseases.


Subject(s)
Air Pollutants , Air Pollution , Adult , Humans , Air Pollutants/toxicity , Air Pollutants/analysis , DNA Methylation , Environmental Exposure/analysis , Air Pollution/adverse effects , Air Pollution/analysis , Particulate Matter/toxicity , Particulate Matter/analysis , Epigenesis, Genetic
17.
Nucleic Acids Res ; 49(5): e29, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33330940

ABSTRACT

Optogenetic control of CRISPR-Cas9 systems has significantly improved our ability to perform genome perturbations in living cells with high precision in time and space. As new Cas orthologues with advantageous properties are rapidly being discovered and engineered, the need for straightforward strategies to control their activity via exogenous stimuli persists. The Cas9 from Neisseria meningitidis (Nme) is a particularly small and target-specific Cas9 orthologue, and thus of high interest for in vivo genome editing applications. Here, we report the first optogenetic tool to control NmeCas9 activity in mammalian cells via an engineered, light-dependent anti-CRISPR (Acr) protein. Building on our previous Acr engineering work, we created hybrids between the NmeCas9 inhibitor AcrIIC3 and the LOV2 blue light sensory domain from Avena sativa. Two AcrIIC3-LOV2 hybrids from our collection potently blocked NmeCas9 activity in the dark, while permitting robust genome editing at various endogenous loci upon blue light irradiation. Structural analysis revealed that, within these hybrids, the LOV2 domain is located in striking proximity to the Cas9 binding surface. Together, our work demonstrates optogenetic regulation of a type II-C CRISPR effector and might suggest a new route for the design of optogenetic Acrs.


Subject(s)
CRISPR-Associated Protein 9/antagonists & inhibitors , CRISPR-Associated Protein 9/chemistry , CRISPR-Cas Systems , Gene Editing/methods , Neisseria meningitidis/enzymology , Optogenetics/methods , Cell Line , HEK293 Cells , Humans , Light , Models, Molecular , Protein Engineering , Proteins/chemistry , Proteins/radiation effects
18.
J Cell Sci ; 133(11)2020 06 01.
Article in English | MEDLINE | ID: mdl-32295847

ABSTRACT

3D cell cultures enable the in vitro study of dynamic biological processes such as the cell cycle, but their use in high-throughput screens remains impractical with conventional fluorescent microscopy. Here, we present a screening workflow for the automated evaluation of mitotic phenotypes in 3D cell cultures by light-sheet microscopy. After sample preparation by a liquid handling robot, cell spheroids are imaged for 24 h in toto with a dual-view inverted selective plane illumination microscope (diSPIM) with a much improved signal-to-noise ratio, higher imaging speed, isotropic resolution and reduced light exposure compared to a spinning disc confocal microscope. A dedicated high-content image processing pipeline implements convolutional neural network-based phenotype classification. We illustrate the potential of our approach using siRNA knockdown and epigenetic modification of 28 mitotic target genes for assessing their phenotypic role in mitosis. By rendering light-sheet microscopy operational for high-throughput screening applications, this workflow enables target gene characterization or drug candidate evaluation in tissue-like 3D cell culture models.


Subject(s)
Image Processing, Computer-Assisted , Spheroids, Cellular , Microscopy, Fluorescence , Mitosis , Phenotype
19.
Gastroenterology ; 160(4): 1330-1344.e11, 2021 03.
Article in English | MEDLINE | ID: mdl-33212097

ABSTRACT

BACKGROUND & AIMS: Molecular evidence of cellular heterogeneity in the human exocrine pancreas has not been yet established because of the local concentration and cascade of hydrolytic enzymes that can rapidly degrade cells and RNA upon pancreatic resection. We sought to better understand the heterogeneity and cellular composition of the pancreas in neonates and adults in healthy and diseased conditions using single-cell sequencing approaches. METHODS: We innovated single-nucleus RNA-sequencing protocols and profiled more than 120,000 cells from pancreata of adult and neonatal human donors. We validated the single-nucleus findings using RNA fluorescence in situ hybridization, in situ sequencing, and computational approaches. RESULTS: We created the first comprehensive atlas of human pancreas cells including epithelial and nonepithelial constituents, and uncovered 3 distinct acinar cell types, with possible implications for homeostatic and inflammatory processes of the pancreas. The comparison with neonatal single-nucleus sequencing data showed a different cellular composition of the endocrine tissue, highlighting the tissue dynamics occurring during development. By applying spatial cartography, involving cell proximity mapping through in situ sequencing, we found evidence of specific cell type neighborhoods, dynamic topographies in the endocrine and exocrine pancreas, and principles of morphologic organization of the organ. Furthermore, similar analyses in chronic pancreatitis biopsy samples showed the presence of acinar-REG+ cells, a reciprocal association between macrophages and activated stellate cells, and a new potential role of tuft cells in this disease. CONCLUSIONS: Our human pancreas cell atlas can be interrogated to understand pancreatic cell biology and provides a crucial reference set for comparisons with diseased tissue samples to map the cellular foundations of pancreatic diseases.


Subject(s)
Cell Nucleus/metabolism , Pancreas, Exocrine/cytology , Adolescent , Adult , Age Factors , Aged , Animals , Cell Fractionation , Child , Child, Preschool , Female , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Middle Aged , Models, Animal , Pancreas, Exocrine/growth & development , Pancreas, Exocrine/metabolism , RNA-Seq , Single-Cell Analysis/methods , Swine , Young Adult
20.
Am J Pathol ; 191(4): 602-617, 2021 04.
Article in English | MEDLINE | ID: mdl-33497701

ABSTRACT

Solitary fibrous tumors (SFTs) harbor recurrent NAB2-STAT6 gene fusions, promoting constitutional up-regulation of oncogenic early growth response 1 (EGR1)-dependent gene expression. SFTs with the most common canonical NAB2 exon 4-STAT6 exon 2 fusion variant are often located in the thorax (pleuropulmonary) and are less cellular with abundant collagen. In contrast, SFTs with NAB2 exon 6-STAT6 exon 16/17 fusion variants typically display a cellular round to ovoid cell morphology and are often located in the deep soft tissue of the retroperitoneum and intra-abdominal pelvic region or in the meninges. Here, we employed next-generation sequencing-based gene expression profiling to identify significant differences in gene expression associated with anatomic localization and NAB2-STAT6 gene fusion variants. SFTs with the NAB2 exon 4-STAT6 exon 2 fusion variant showed a transcriptional signature enriched for genes involved in DNA binding, gene transcription, and nuclear localization, whereas SFTs with the NAB2 exon 6-STAT6 exon 16/17 fusion variants were enriched for genes involved in tyrosine kinase signaling, cell proliferation, and cytoplasmic localization. Specific transcription factor binding motifs were enriched among differentially expressed genes in SFTs with different fusion variants, implicating co-transcription factors in the modification of chimeric NGFI-A binding protein 2 (NAB2)-STAT6-dependent deregulation of EGR1-dependent gene expression. In summary, this study establishes a potential molecular biologic basis for clinicopathologic differences in SFTs with distinct NAB2-STAT6 gene fusion variants.


Subject(s)
Biomarkers, Tumor/genetics , Repressor Proteins/genetics , STAT6 Transcription Factor/genetics , Solitary Fibrous Tumors/genetics , Exons/genetics , Female , Gene Expression/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Oncogene Proteins, Fusion/genetics , Repressor Proteins/metabolism , Solitary Fibrous Tumors/pathology
SELECTION OF CITATIONS
SEARCH DETAIL